Skip to content

This report is written by MaltSci based on the latest literature and research findings


What are the mechanisms of viral immune evasion?

Abstract

The ability of viruses to evade the host immune system is a critical factor in their pathogenicity and persistence, posing significant challenges for understanding viral biology and developing effective vaccines and antiviral therapies. This review explores the sophisticated mechanisms employed by viruses to escape detection and destruction by the immune system, categorized into innate and adaptive immune evasion strategies. Key tactics include alterations in viral antigens to prevent antibody recognition, inhibition of interferon signaling pathways, and disruption of antigen presentation mechanisms that thwart T cell responses. Notable case studies, including HIV, Influenza, and SARS-CoV-2, illustrate the diverse strategies of immune evasion. For instance, HIV's rapid mutation rate allows it to escape T cell recognition, while Influenza employs the non-structural protein 1 (NS1) to inhibit type I interferon signaling. SARS-CoV-2 downregulates MHC-I expression and induces hyper-inflammatory responses, complicating immune recognition. The insights gained from understanding these mechanisms are essential for informing vaccine development strategies and guiding the design of novel antiviral therapies. As viral infections continue to pose significant public health threats, particularly in the wake of emerging viruses, ongoing research is crucial to enhance host defenses and combat viral infections effectively.

Outline

This report will discuss the following questions.

  • 1 Introduction
  • 2 Mechanisms of Viral Immune Evasion
    • 2.1 Alteration of Viral Antigens
    • 2.2 Inhibition of Interferon Signaling
    • 2.3 Disruption of Antigen Presentation
    • 2.4 Modulation of Apoptotic Pathways
  • 3 Innate Immune Evasion Strategies
    • 3.1 Evading Pattern Recognition Receptors
    • 3.2 Counteracting Complement Activation
  • 4 Adaptive Immune Evasion Strategies
    • 4.1 T Cell Evasion Mechanisms
    • 4.2 B Cell Response Modulation
  • 5 Case Studies of Viral Immune Evasion
    • 5.1 HIV
    • 5.2 Influenza Virus
    • 5.3 SARS-CoV-2
  • 6 Implications for Vaccine Development
  • 7 Conclusion

1 Introduction

The ability of viruses to evade the host immune system is a crucial factor in their pathogenicity and persistence. This phenomenon, known as viral immune evasion, represents a significant challenge for both our understanding of viral biology and the development of effective vaccines and antiviral therapies. Viruses have evolved a myriad of sophisticated strategies to escape detection and destruction by the immune system, employing mechanisms that disrupt innate and adaptive immune responses. Understanding these mechanisms is not only vital for elucidating the dynamics of viral infections but also for designing innovative therapeutic interventions that can enhance host defenses.

Research in the field has demonstrated that viruses utilize various tactics to avoid immune recognition. These include alterations in viral antigens, inhibition of immune signaling pathways, and manipulation of host cellular responses [1][2]. For instance, some viruses can change their surface proteins to prevent recognition by antibodies, while others interfere with the interferon signaling pathways that are critical for mounting an antiviral response [1][3]. Additionally, viruses can disrupt antigen presentation mechanisms, thereby preventing T cells from recognizing and eliminating infected cells [1][4].

The significance of studying viral immune evasion mechanisms cannot be overstated. As viral infections continue to pose significant public health threats, particularly in the wake of emerging viruses such as SARS-CoV-2, a deeper understanding of how these pathogens manipulate host defenses is essential [2]. The insights gained from such studies can inform vaccine development strategies and guide the design of novel antiviral therapies that target specific evasion mechanisms [1][5].

Current research has identified several key mechanisms through which viruses evade the immune system. These mechanisms can be categorized into innate and adaptive immune evasion strategies. Innate immune evasion includes tactics such as evading pattern recognition receptors and counteracting complement activation [6][7]. Adaptive immune evasion, on the other hand, encompasses strategies that target T cell and B cell responses, effectively subverting the host's acquired immunity [8][9].

This review is organized as follows: we will first explore the various mechanisms of viral immune evasion, detailing how viruses alter their antigens, inhibit interferon signaling, disrupt antigen presentation, and modulate apoptotic pathways. Next, we will examine specific strategies employed by viruses to evade innate immunity, including the evasion of pattern recognition receptors and the counteraction of complement activation. Following this, we will discuss adaptive immune evasion strategies, focusing on T cell evasion mechanisms and the modulation of B cell responses. We will then present case studies of notable viruses, including HIV, Influenza, and SARS-CoV-2, to illustrate the diverse strategies of immune evasion in action. Finally, we will discuss the implications of these findings for vaccine development and therapeutic interventions, emphasizing the need for ongoing research to combat viral infections effectively.

In summary, understanding the mechanisms of viral immune evasion is critical for advancing our knowledge of viral pathogenesis and for developing effective strategies to enhance host immunity. The complex interplay between viruses and the immune system underscores the necessity for continued research in this vital area of biomedical science.

2 Mechanisms of Viral Immune Evasion

2.1 Alteration of Viral Antigens

Viral immune evasion is a sophisticated and multifaceted process that allows viruses to persist in the host despite the immune system's efforts to eliminate them. One of the primary mechanisms employed by viruses to evade immune detection is the alteration of viral antigens, which can significantly affect the host's immune response.

Viruses exhibit remarkable plasticity in their surface proteins, which are crucial for their recognition by the immune system. For instance, influenza viruses are known for their ability to escape the human antibody response by altering the amino acids in their surface proteins, particularly in hypervariable domains. This structural plasticity allows the virus to continuously change its antigenic profile, making it difficult for the immune system to recognize and mount an effective response against previously encountered strains (Crow 2012) [10].

Similarly, SARS-CoV-2, the virus responsible for COVID-19, has developed strategies to evade antibody neutralization through mutations in its spike protein. These mutations not only enhance the virus's ability to infect cells but also impair the immune system's ability to recognize and neutralize the virus. Specifically, SARS-CoV-2 proteins, such as ORF 6 and ORF 8, have been shown to downregulate MHC-I molecule expression, thereby reducing the effectiveness of cytotoxic T-cell responses (Rubio-Casillas et al. 2022) [2].

In addition to direct alterations of viral antigens, viruses can also manipulate host immune responses through various means. For example, they can interfere with antigen processing and presentation pathways, thereby preventing the effective recognition of infected cells by T cells. This interference often involves the modulation of host cytokine networks, which can create an immunosuppressive environment that further facilitates viral persistence (Eslami et al. 2025) [11].

Moreover, some viruses exploit mechanisms such as exosome-mediated communication to enhance their survival. By incorporating viral components into exosomes, viruses can cloak their antigens, effectively decoying the immune system and increasing their chances of evading detection (Anderson et al. 2016) [12].

The continuous evolution of viral antigens through mechanisms such as antigenic variation, as well as the strategic manipulation of host immune pathways, underscores the complex interplay between viruses and the immune system. Understanding these mechanisms is critical for developing effective antiviral therapies and vaccines that can counteract the sophisticated strategies employed by viruses to evade immune surveillance.

2.2 Inhibition of Interferon Signaling

Viral immune evasion is a critical factor in the ability of viruses to establish and maintain infection within a host. One of the primary strategies employed by viruses to achieve this is the inhibition of interferon (IFN) signaling, which is a crucial component of the host's antiviral response. The mechanisms by which viruses inhibit IFN signaling are diverse and sophisticated, allowing them to effectively counteract the host's immune defenses.

Firstly, viruses can prevent the initial detection of viral components by the host's immune system. This is achieved through the disruption of pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)-like receptors. By hindering the recognition of viral RNA or DNA, viruses can evade the triggering of the IFN response that would normally occur upon infection (Taylor & Mossman, 2013) [13].

Secondly, some viruses actively inhibit the production of type I IFNs. This can occur through the degradation or disarming of transcription factors essential for IFN gene expression, such as interferon regulatory factors (IRFs) and nuclear factor-κB (NF-κB). For instance, certain viral proteins have been shown to interfere with the phosphorylation and nuclear accumulation of IRF-7, which is critical for the induction of IFN (Zhu et al., 2002) [14]. Moreover, viruses can induce a general block on host cell transcription, thereby preventing the production of IFNs altogether (Schulz & Mossman, 2016) [15].

Additionally, once IFNs are produced, viruses can interfere with the signaling pathways activated by these cytokines. This is often accomplished by targeting the type I IFN receptor and disrupting the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway. By impeding this signaling cascade, viruses can inhibit the downstream effects of IFNs, including the activation of IFN-stimulated genes (ISGs) that play a vital role in establishing an antiviral state (Wang et al., 2018) [16].

Viruses also utilize more complex mechanisms to evade the IFN response. For example, they can co-opt cellular negative regulatory systems to their advantage, effectively using host pathways to inhibit the immune response. Some viruses produce proteins that bind to and sequester key components of the IFN signaling pathway, rendering them inactive (Devasthanam, 2014) [17].

Finally, recent studies have highlighted the role of viral non-structural proteins in suppressing IFN production. These proteins can inhibit the signaling pathways that lead to IFN production and block the expression of ISGs, further enhancing viral replication and persistence within the host (Chiang & Liu, 2018) [18].

In summary, the mechanisms of viral immune evasion, particularly through the inhibition of interferon signaling, involve a multifaceted approach. Viruses employ strategies to prevent detection by PRRs, inhibit IFN production and signaling, and utilize host regulatory pathways to subvert the immune response. Understanding these mechanisms is crucial for the development of effective antiviral therapies and vaccines.

2.3 Disruption of Antigen Presentation

Viral immune evasion is a critical aspect of viral pathogenesis, enabling viruses to persist within the host and evade immune surveillance. One of the primary mechanisms employed by viruses to achieve this is the disruption of antigen presentation, particularly through the inhibition of the major histocompatibility complex class I (MHC-I) pathway. This disruption has profound implications for the host's ability to mount an effective immune response.

Viruses utilize various strategies to interfere with the MHC-I antigen presentation pathway. For instance, certain viral proteins have been shown to downregulate the expression of MHC-I molecules on the surface of infected cells. This is exemplified by SARS-CoV-2, where the open reading frames (ORF) 6 and 8 produce viral proteins that specifically target and reduce MHC-I expression, thereby preventing the presentation of viral peptides to CD8+ cytotoxic T lymphocytes (CTLs) (Rubio-Casillas et al. 2022) [2]. Similarly, chronic viral infections such as those caused by HIV, HBV, and HCV also involve the downregulation of MHC-I molecules, disrupting the host's ability to recognize and eliminate infected cells (Eslami et al. 2025) [11].

Moreover, viruses may employ additional tactics to interfere with the intracellular processing of antigens. For example, some viral proteins can inhibit the transport of peptides to the endoplasmic reticulum, where MHC-I molecules are loaded with antigenic peptides. This interference limits the availability of peptides that would normally be presented to CTLs, thereby diminishing the immune response (Hansen & Bouvier 2009) [19].

The consequences of such disruptions are significant. By preventing the effective presentation of viral antigens, viruses can remain "invisible" to the immune system, particularly during the latency phase of infection. This is particularly evident in certain herpesviruses, where intrinsic, genome-encoded elements actively interfere with antigen processing and display (Wan et al. 2025) [20]. These mechanisms of primary sequence intrinsic immune evasion (PSI) not only limit antigen production but also contribute to the persistence of the virus in the host.

In summary, the disruption of antigen presentation is a multifaceted strategy employed by viruses to evade immune detection. By downregulating MHC-I molecules and interfering with antigen processing and presentation, viruses can effectively evade cytotoxic T-cell responses, thereby facilitating persistent infections and complicating vaccine development and antiviral strategies. Understanding these mechanisms is essential for the design of effective therapeutic interventions aimed at enhancing immune recognition and clearance of viral infections.

2.4 Modulation of Apoptotic Pathways

Viruses have evolved a variety of mechanisms to evade the host immune system, and one critical strategy involves the modulation of apoptotic pathways. Apoptosis, or programmed cell death, serves as a primary defense mechanism against viral infections, enabling the elimination of infected cells. However, many viruses have developed sophisticated tactics to manipulate these pathways to their advantage, thereby enhancing their survival and replication.

One of the fundamental mechanisms by which viruses evade apoptosis is through the direct inhibition of apoptotic signaling pathways. This includes the encoding of viral proteins that either inhibit pro-apoptotic factors or activate anti-apoptotic pathways. For instance, some viruses produce proteins that are homologous to host-derived apoptosis-regulatory proteins, such as members of the Bcl-2 family, which are known to inhibit the apoptotic process [21]. By mimicking these host proteins, viruses can effectively block the apoptotic signals that would typically lead to cell death.

Additionally, viruses can modulate the host's apoptotic machinery by interacting with cell surface receptors or components of the apoptotic pathways. For example, Human Immunodeficiency Virus (HIV) has been shown to alter normal apoptotic signaling, resulting in increased viral load and the establishment of viral reservoirs, which ultimately enhances infectivity [22]. The interactions of viral proteins with the host's pro- and anti-apoptotic responses play a crucial role in this dynamic, allowing the virus to manipulate the host cell environment to favor its own replication.

Furthermore, certain viruses may exploit the apoptotic machinery to eliminate uninfected immune cells, thereby reducing the overall immune response against the infection. This is particularly evident in the case of HIV, which activates apoptotic programs that destroy immune effectors, contributing to the chronic immune activation observed in infected individuals [23]. By doing so, HIV not only protects itself from the immune attack but also creates a more favorable environment for its propagation.

Moreover, the suppression of apoptosis is not limited to RNA viruses like HIV; DNA viruses, such as α-herpesviruses, have also developed multifaceted strategies to inhibit programmed cell death. These viruses encode anti-apoptotic factors that interfere with both the death receptor pathway and the mitochondria-mediated pathway of apoptosis, promoting their own replication and evasion of host defenses [24].

In summary, the mechanisms of viral immune evasion through the modulation of apoptotic pathways include the direct inhibition of apoptotic signals, manipulation of host cell responses, and the strategic activation of apoptosis in immune cells to diminish the host's defense. These tactics not only facilitate viral survival and replication but also highlight the complex interplay between viruses and the host immune system in the context of infection [21][22][25].

3 Innate Immune Evasion Strategies

3.1 Evading Pattern Recognition Receptors

Viral immune evasion is a critical aspect of viral pathogenesis, particularly in the context of the innate immune system, which serves as the first line of defense against viral infections. A central component of this system is the pattern recognition receptors (PRRs), which detect viral components and initiate antiviral responses. Viruses have evolved various strategies to evade detection by these receptors, thereby facilitating their replication and persistence within the host.

One prominent mechanism employed by viruses to evade PRRs involves the modification or sequestration of viral nucleic acids. For instance, viruses may alter their RNA or DNA to prevent recognition by intracellular sensors such as RIG-I-like receptors (RLRs) and cyclic GMP-AMP synthase (cGAS). This alteration can include the addition of caps or other modifications that inhibit the binding of PRRs, effectively masking the viral genome from immune detection [26].

Another strategy involves the interference with the signaling pathways activated by PRRs. Many viruses disrupt key signaling cascades, such as the nuclear factor kappa-B (NF-κB) pathway and the Janus kinase-signal transducers and activators of transcription (JAK-STAT) pathway, which are crucial for the induction of type I interferons (IFNs) and other pro-inflammatory cytokines. For example, enteroviruses utilize specific proteases, such as 2A and 3C proteases, to cleave host proteins that are integral to immune signaling, thereby inhibiting the host's antiviral response [27].

Moreover, some viruses can directly target PRRs themselves, leading to their degradation or functional inhibition. This includes the cleavage of PRRs or their adaptor proteins, which prevents the downstream signaling necessary for an effective immune response [28]. For instance, the human immunodeficiency virus (HIV-1) has been shown to exploit host proteins to evade DNA sensing mechanisms, particularly through the action of TREX1, a host exonuclease that plays a role in the recognition of viral DNA [28].

In addition to direct targeting, viruses can also manipulate the cellular environment to create conditions that are unfavorable for PRR activation. This includes the sequestration or relocalization of PRRs away from their sites of action, thus preventing them from engaging with viral components [29]. The ability of viruses to modulate these pathways underscores their evolutionary adaptation to circumvent host defenses.

The complexity of viral immune evasion mechanisms illustrates a dynamic interplay between viruses and the host immune system. Understanding these mechanisms at a molecular level is crucial for developing effective vaccines and antiviral therapies, as it can provide insights into potential targets for intervention [30].

In summary, viral evasion of innate immunity, particularly through the evasion of PRRs, involves a multifaceted approach that includes modification of viral nucleic acids, disruption of immune signaling pathways, direct targeting of PRRs, and manipulation of the host cellular environment. These strategies collectively enhance viral replication and persistence, posing significant challenges for the host immune response.

3.2 Counteracting Complement Activation

Viruses have evolved a range of sophisticated mechanisms to evade the host's immune responses, particularly targeting the complement system, which is a crucial component of innate immunity. The complement system operates through three primary pathways: the classical, alternative, and lectin pathways, leading to the opsonization of pathogens, inflammation, and cell lysis. The ability of viruses to counteract complement activation is essential for their survival and propagation within the host.

One significant strategy employed by various viruses is the inhibition of complement activation. For instance, many viruses produce proteins that can directly interfere with complement components, thereby preventing the formation of the C3 convertase, which is critical for downstream complement activation. This strategy is vital as complement activation not only leads to the opsonization of viral particles but also triggers inflammatory responses that can eliminate the virus.

Moreover, viruses have been shown to exploit complement regulatory mechanisms to their advantage. They can mimic host proteins or utilize viral proteins that resemble complement regulators to inhibit complement activation. For example, the complement evasion strategies discussed by Favoreel et al. (2003) highlight how certain viruses can block complement activation pathways, thus avoiding recognition and clearance by the immune system[31].

Another mechanism involves the use of the complement system to enhance viral infectivity. Some viruses can bind to complement components or receptors, which may facilitate their entry into host cells or help them establish a latent infection. This interaction not only aids in evading the immune response but can also manipulate the immune system to create an environment conducive to viral replication and persistence[32].

Additionally, specific viruses have developed mechanisms to modulate the inflammatory responses triggered by complement activation. By doing so, they can reduce the effectiveness of the immune response while simultaneously promoting their own replication and spread. This manipulation of the host's immune response is a common theme among many viral pathogens, including those causing significant diseases[33].

In summary, the mechanisms of viral immune evasion through counteracting complement activation are multifaceted. They include direct inhibition of complement components, exploitation of complement regulatory proteins, and manipulation of the inflammatory responses associated with complement activation. Understanding these strategies is crucial for developing effective vaccines and therapeutic interventions against viral infections[34][35].

4 Adaptive Immune Evasion Strategies

4.1 T Cell Evasion Mechanisms

Viral immune evasion represents a significant challenge in the context of adaptive immunity, particularly concerning T cell responses. The mechanisms through which viruses evade T cell immunity are diverse and complex, often involving sophisticated strategies to undermine the host's immune recognition and response capabilities.

One primary mechanism of T cell evasion is the alteration of viral epitopes. Viruses can accumulate mutations in T cell epitopes, which leads to the loss of recognition by virus-specific CD8+ T cells. This phenomenon has been well-documented in the context of HIV-1, where the virus employs non-synonymous mutations to escape detection by the immune system, resulting in a significant loss of T cell recognition and a consequent failure to control the infection [36].

Another notable strategy is the manipulation of the major histocompatibility complex (MHC) class I antigen presentation pathway. Many viruses have evolved proteins that interfere with the presentation of viral peptides on MHC class I molecules, thereby preventing CD8+ T cells from recognizing and eliminating infected cells. This form of immune evasion has been extensively studied in herpesviruses, which utilize various mechanisms to disrupt the normal processing and presentation of antigens [37].

Additionally, T cell exhaustion is a critical factor in the context of chronic viral infections. During persistent infections, T cells can enter a state of dysfunction characterized by reduced proliferation and effector function, which is often driven by sustained antigen exposure. This exhaustion can lead to an impaired ability to control viral replication [38]. Mechanistically, T cell exhaustion is associated with metabolic dysfunction, where the altered metabolic pathways in T cells limit their effectiveness in combating chronic infections [38].

Moreover, some viruses employ strategies that promote T cell exhaustion through the upregulation of inhibitory receptors on T cells, such as PD-1, which further dampens T cell responses. This strategy is particularly relevant in the context of HIV and hepatitis viruses, where the virus exploits the immune system's regulatory pathways to evade effective T cell-mediated responses [39].

Lastly, viral recombination is another mechanism through which T cells may be evaded. In HIV-1 infections, recombination between different viral strains can lead to the emergence of variants that escape T cell recognition, further complicating the host's ability to mount an effective immune response [36].

In summary, the mechanisms of T cell evasion by viruses are multifaceted, involving mutations in viral epitopes, manipulation of antigen presentation pathways, induction of T cell exhaustion, and the use of recombination to generate escape variants. Understanding these mechanisms is crucial for developing effective therapeutic strategies to enhance T cell responses against viral infections.

4.2 B Cell Response Modulation

Viral immune evasion is a critical aspect of viral pathogenesis, particularly concerning how viruses circumvent host immune responses to establish persistent infections. A significant area of focus is the modulation of B cell responses, which is essential for the production of antibodies that can neutralize viral pathogens.

Viruses employ several mechanisms to interfere with B cell responses. One prominent strategy is the alteration of the host cytokine milieu, which can influence B cell activation and differentiation. By modulating cytokine levels, viruses can promote the development of immunoregulatory cells that suppress effective B cell responses, thereby allowing the virus to persist in the host [40].

Moreover, viruses can induce the production of non-neutralizing antibodies that interfere with the activity of neutralizing antibodies (nAbs). This phenomenon, referred to as "neutralization interfering antibodies," can diminish the efficacy of the immune response, facilitating viral escape from neutralization [41]. Such mechanisms are particularly problematic in chronic infections, where robust humoral responses do not translate into effective viral clearance [41].

Additionally, viruses can exploit mechanisms related to the modulation of B cell immunodominance. This is the phenomenon where the immune response is skewed towards certain antigenic determinants, potentially neglecting others that may be critical for effective viral neutralization. By altering the expression of viral antigens or presenting them in a manner that shifts the focus of the immune response, viruses can evade recognition by B cells [42].

In the context of hepatitis C virus (HCV), studies have identified specific mutations that allow the virus to evade neutralizing antibodies by altering the use of host-cell entry factors. These mutations can affect virus-antibody interactions, leading to persistent infection [43]. This highlights the dynamic interplay between viral evolution and host immune responses, emphasizing the need for continuous adaptation by both the virus and the immune system.

Understanding these mechanisms is crucial for developing novel antiviral therapies and vaccines. By elucidating how viruses modulate B cell responses, researchers can design strategies that enhance the effectiveness of the immune response against viral infections, potentially leading to improved outcomes in managing chronic viral diseases [44].

5 Case Studies of Viral Immune Evasion

5.1 HIV

Human Immunodeficiency Virus (HIV) employs a multitude of sophisticated mechanisms to evade the host immune system, significantly complicating efforts to develop effective vaccines and therapeutic strategies. These mechanisms can be categorized into various strategies that target both the innate and adaptive immune responses.

  1. Genomic Mutations and Antigenic Variation: HIV is characterized by a high mutation rate, which allows it to rapidly alter its surface proteins. This is particularly evident in the envelope glycoprotein (Env), which is the primary target for neutralizing antibodies. HIV utilizes conformational changes in Env to present different antigenic states to the immune system, effectively hiding vulnerable epitopes from recognition. For instance, studies have shown that the Env glycoprotein can adopt conformations that are less recognizable to the immune system, thus allowing the virus to persist in the presence of robust antibody responses [45].

  2. Downregulation of Major Histocompatibility Complex (MHC) Molecules: HIV has developed mechanisms to downregulate MHC class I molecules on the surface of infected cells. This is achieved through viral proteins such as Nef, which mediates the internalization of MHC-I molecules from the cell surface, thereby reducing the visibility of infected cells to CD8+ T cells [46]. Additionally, the Tat protein can repress MHC-I transcription, further impairing the ability of the immune system to recognize and eliminate infected cells [46].

  3. Formation of Latent Viral Reservoirs: HIV can establish latent reservoirs in the host, where the virus remains dormant within infected cells, evading immune detection. These reservoirs can reactivate under certain conditions, leading to renewed viral replication and disease progression [47].

  4. Interference with Innate Immune Responses: HIV exploits various host factors to evade detection by the innate immune system. For example, it can suppress the activity of restriction factors such as APOBEC3G and TRIM5α, which are part of the intrinsic immune defense against viral infections [48]. Furthermore, HIV can manipulate host proteins involved in DNA sensing, such as TREX1, to evade recognition by the immune system [28].

  5. Exploitation of Immune Cell Mechanisms: HIV can induce apoptosis in immune cells, particularly CD4+ T cells, thereby depleting a critical component of the adaptive immune response. This strategy not only reduces the number of immune cells available to combat the infection but also contributes to a state of chronic immune activation, which can lead to immune exhaustion [23].

  6. Natural Killer (NK) Cell Evasion: HIV can escape NK cell-mediated immune pressure by mutating specific epitopes presented by MHC class I molecules. For instance, certain mutations in the p24 Gag protein were found to enhance binding to inhibitory receptors on NK cells, thereby diminishing the activation of these immune cells against HIV-infected targets [49].

  7. Induction of Immune Dysfunction: HIV infection leads to a broad dysfunction of the immune system, including clonal expansion and excessive activation of T cells, resulting in immunosenescence and exhaustion [50]. This dysfunction impairs the overall ability of the immune system to mount effective responses against the virus.

Understanding these mechanisms of immune evasion is crucial for the development of novel therapeutic and vaccine strategies aimed at overcoming the challenges posed by HIV. Insights into how HIV manipulates the immune response can guide researchers in designing interventions that enhance immune recognition and clearance of the virus.

5.2 Influenza Virus

Influenza viruses exhibit a variety of mechanisms to evade host immune responses, which is critical for their survival and replication. These mechanisms involve both the direct manipulation of host cellular processes and the alteration of immune signaling pathways.

One prominent mechanism of immune evasion by the influenza virus is the function of the non-structural protein 1 (NS1). NS1 inhibits type I interferon (IFN) signaling pathways, which are essential for antiviral responses. In a study, it was shown that mice infected with an NS1-expressing influenza A H1N1 virus exhibited lower expression levels of IFNβ in the lungs. Additionally, human alveolar epithelial A549 cells infected with the H1N1 virus produced antiviral IFNβ, but transfection with H1N1 NS1 resulted in the loss of this ability, indicating that NS1 interferes with both the production of IFNβ and the expression of IFN receptors. Furthermore, NS1 inhibits the activation of signal transducers and activators of transcription (STAT)1 and STAT2, which are crucial for the downstream signaling of IFNβ production, thus playing a significant role in the virus's evasion strategy [51].

Another mechanism involves host shutoff, where the influenza virus down-regulates host gene expression to take control of the infected cells. This process is facilitated by several viral proteins, including NS1, the RNA-dependent RNA polymerase, and the endoribonuclease PA-X. These proteins contribute to the alteration of translation, RNA synthesis, and stability, thereby hindering the host's immune response and facilitating viral replication [52].

Additionally, influenza viruses employ strategies to evade natural killer (NK) cell recognition. The neuraminidase (NA) protein plays a crucial role in this process by removing sialic acid residues from the NKp46 receptor on NK cells, which diminishes the recognition of hemagglutinin (HA) on infected cells. This mechanism highlights how the virus can interfere with innate immune responses, particularly those mediated by NK cells, thereby enhancing its survival [53].

The virus also activates various pathways that suppress host innate immunity. For instance, the epidermal growth factor receptor (EGFR) and extracellular signal-regulated kinase (ERK) signaling pathways are activated upon influenza A virus infection, leading to the suppression of type I and III IFNs and interferon-stimulated genes (ISGs). This activation of EGFR/ERK signaling is facilitated by the protein tyrosine phosphatase SHP2, which plays a critical role in modulating antiviral responses [54].

Moreover, influenza viruses can induce host shut-off mechanisms that disrupt the production of type I IFNs. The viruses exploit various host proteins involved in the IFN signaling pathway, such as tripartite motif containing 25 (TRIM25) and mitochondrial antiviral signaling protein (MAVS), to evade detection and inhibit effective immune responses [55].

In summary, influenza viruses utilize multiple strategies to evade host immune responses, including the inhibition of IFN signaling through NS1, host shutoff mechanisms, alteration of NK cell recognition via neuraminidase, and the activation of signaling pathways that suppress antiviral responses. These mechanisms collectively enhance the virus's ability to replicate and persist within the host.

5.3 SARS-CoV-2

SARS-CoV-2 employs multiple sophisticated mechanisms to evade the host's immune system, which are critical for its pathogenesis and the persistence of infection. The following outlines key strategies identified in recent studies.

One primary mechanism is the suppression of type I interferon (IFN) production, which is crucial for initiating the innate immune response. SARS-CoV-2 achieves this through various non-structural proteins (NSPs) and accessory proteins that interfere with the signaling pathways involved in IFN production. For instance, NSP2 has been shown to repress the translation of IFN-β mRNA by interacting with the cellular protein GIGYF2, which enhances the binding of GIGYF2 to the mRNA cap-binding protein 4EHP, leading to reduced IFN-β synthesis [56]. Additionally, the virus can downregulate the expression of major histocompatibility complex class I (MHC-I) molecules through proteins such as ORF6 and ORF8, thus hindering the recognition of infected cells by cytotoxic T lymphocytes [2].

SARS-CoV-2 also targets the innate immune cells directly. It can exhaust natural killer (NK) cell-mediated cytotoxicity by downregulating stress ligands recognized by the NKG2D receptor on NK cells [57]. Furthermore, the virus can impair the activation of pattern recognition receptors (PRRs) that are essential for the detection of viral components, thereby compromising the signaling pathways that lead to the activation of immune responses [58].

Another significant mechanism of immune evasion involves the induction of a hyper-inflammatory response, known as a cytokine storm, which can lead to severe tissue damage and further complications in COVID-19 patients. SARS-CoV-2 triggers the overactivation of the NLRP3 inflammasome, resulting in the excessive release of pro-inflammatory cytokines [59]. This dysregulation of the immune response not only allows the virus to persist but also contributes to the pathogenesis of severe disease [60].

The virus's ability to generate extracellular vesicles (EVs) has also been implicated in immune evasion. These EVs can facilitate cell-to-cell transmission of the virus while evading neutralizing antibodies, thereby promoting viral spread without triggering an effective immune response [61].

Additionally, SARS-CoV-2 can exploit host cellular pathways to its advantage. For example, it can hijack cellular machinery involved in protein synthesis and metabolism to favor its replication while simultaneously inhibiting the host's antiviral responses [62].

Overall, the intricate mechanisms of immune evasion employed by SARS-CoV-2 highlight the challenges in developing effective therapeutic strategies and vaccines against this virus. Understanding these mechanisms is crucial for advancing antiviral therapies and improving pandemic preparedness [63][64].

6 Implications for Vaccine Development

7 Conclusion

The mechanisms of viral immune evasion are intricate and multifaceted, significantly complicating our understanding of viral pathogenesis and the development of effective therapeutic strategies. This review highlights key strategies employed by various viruses, including the alteration of viral antigens, inhibition of interferon signaling, disruption of antigen presentation, and modulation of apoptotic pathways. Notably, viruses like HIV, Influenza, and SARS-CoV-2 exemplify how these evasion tactics can undermine both innate and adaptive immune responses. The continuous evolution of viral strains further exacerbates the challenge of vaccine development, necessitating ongoing research to elucidate these mechanisms. Future studies should focus on identifying novel therapeutic targets that can enhance immune recognition and clearance of viral infections. Moreover, integrating insights from viral immune evasion mechanisms into vaccine design could pave the way for more effective interventions against persistent viral diseases, ultimately improving public health outcomes.

References

  • [1] Marni S Crow;Krystal K Lum;Xinlei Sheng;Bokai Song;Ileana M Cristea. Diverse mechanisms evolved by DNA viruses to inhibit early host defenses.. Critical reviews in biochemistry and molecular biology(IF=6.4). 2016. PMID:27650455. DOI: 10.1080/10409238.2016.1226250.
  • [2] Alberto Rubio-Casillas;Elrashdy M Redwan;Vladimir N Uversky. SARS-CoV-2: A Master of Immune Evasion.. Biomedicines(IF=3.9). 2022. PMID:35740361. DOI: 10.3390/biomedicines10061339.
  • [3] Raquel Muñoz-Moreno;Carles Martínez-Romero;Adolfo García-Sastre. Induction and Evasion of Type-I Interferon Responses during Influenza A Virus Infection.. Cold Spring Harbor perspectives in medicine(IF=10.1). 2021. PMID:32661015. DOI: 10.1101/cshperspect.a038414.
  • [4] T A Banks;B T Rouse. Herpesviruses--immune escape artists?. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America(IF=7.3). 1992. PMID:1315587. DOI: 10.1093/clinids/14.4.933.
  • [5] R J Mrsny. Adjuvants and delivery issues related to immunization: a survey of the recent patent literature.. Journal of drug targeting(IF=3.9). 1998. PMID:9894692. DOI: 10.3109/10611869808996832.
  • [6] Sun-Hwa Lee;Jae U Jung;Robert E Means. 'Complementing' viral infection: mechanisms for evading innate immunity.. Trends in microbiology(IF=14.9). 2003. PMID:14557025. DOI: 10.1016/j.tim.2003.08.004.
  • [7] Yingjie Xiang;Chunxiao Mou;Xing Zhao;Chen Zhuo;Kaichuang Shi;Yun Young Go;Zhenhai Chen. PRRSV GP2a blocks the RLR signaling pathway by targeting RIG-I.. Journal of virology(IF=3.8). 2025. PMID:41070967. DOI: 10.1128/jvi.01135-25.
  • [8] Daniëlle Horst;Marieke C Verweij;Andrew J Davison;Maaike E Ressing;Emmanuel J H J Wiertz. Viral evasion of T cell immunity: ancient mechanisms offering new applications.. Current opinion in immunology(IF=5.8). 2011. PMID:21146386. DOI: 10.1016/j.coi.2010.11.005.
  • [9] Tal Vider-Shalit;Ronit Sarid;Kobi Maman;Lea Tsaban;Ran Levi;Yoram Louzoun. Viruses selectively mutate their CD8+ T-cell epitopes--a large-scale immunomic analysis.. Bioinformatics (Oxford, England)(IF=5.4). 2009. PMID:19478014. DOI: 10.1093/bioinformatics/btp221.
  • [10] James E Crowe. Influenza virus resistance to human neutralizing antibodies.. mBio(IF=4.7). 2012. PMID:22851660. DOI: .
  • [11] Majid Eslami;Neda Arjmand;Fatemeh Mahmoudian;Ali Babaeizad;Hamed Tahmasebi;Fahimeh Fattahi;Valentyn Oksenych. Deciphering Host-Virus Interactions and Advancing Therapeutics for Chronic Viral Infection.. Viruses(IF=3.5). 2025. PMID:40143318. DOI: 10.3390/v17030390.
  • [12] Monique R Anderson;Fatah Kashanchi;Steven Jacobson. Exosomes in Viral Disease.. Neurotherapeutics : the journal of the American Society for Experimental NeuroTherapeutics(IF=6.9). 2016. PMID:27324390. DOI: 10.1007/s13311-016-0450-6.
  • [13] Kathryne E Taylor;Karen L Mossman. Recent advances in understanding viral evasion of type I interferon.. Immunology(IF=5.0). 2013. PMID:23173987. DOI: 10.1111/imm.12038.
  • [14] Fan Xiu Zhu;Sonya M King;Eric J Smith;David E Levy;Yan Yuan. A Kaposi's sarcoma-associated herpesviral protein inhibits virus-mediated induction of type I interferon by blocking IRF-7 phosphorylation and nuclear accumulation.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 2002. PMID:11943871. DOI: 10.1073/pnas.082420599.
  • [15] Katharina S Schulz;Karen L Mossman. Viral Evasion Strategies in Type I IFN Signaling - A Summary of Recent Developments.. Frontiers in immunology(IF=5.9). 2016. PMID:27891131. DOI: 10.3389/fimmu.2016.00498.
  • [16] Yining Wang;Lina Ma;Laszlo Stipkovits;Susan Szathmary;Xuerui Li;Yongsheng Liu. The Strategy of Picornavirus Evading Host Antiviral Responses: Non-structural Proteins Suppress the Production of IFNs.. Frontiers in microbiology(IF=4.5). 2018. PMID:30619109. DOI: 10.3389/fmicb.2018.02943.
  • [17] Anand S Devasthanam. Mechanisms underlying the inhibition of interferon signaling by viruses.. Virulence(IF=5.4). 2014. PMID:24504013. DOI: 10.4161/viru.27902.
  • [18] Hao-Sen Chiang;Helene Minyi Liu. The Molecular Basis of Viral Inhibition of IRF- and STAT-Dependent Immune Responses.. Frontiers in immunology(IF=5.9). 2018. PMID:30671058. DOI: 10.3389/fimmu.2018.03086.
  • [19] Ted H Hansen;Marlene Bouvier. MHC class I antigen presentation: learning from viral evasion strategies.. Nature reviews. Immunology(IF=60.9). 2009. PMID:19498380. DOI: 10.1038/nri2575.
  • [20] Li Wan;Masahiro Shuda;Yuan Chang;Patrick S Moore. Primary Sequence-Intrinsic Immune Evasion by Viral Proteins Guides CTL-Based Vaccine Strategies.. Viruses(IF=3.5). 2025. PMID:40872750. DOI: 10.3390/v17081035.
  • [21] Lorenzo Galluzzi;Catherine Brenner;Eugenia Morselli;Zahia Touat;Guido Kroemer. Viral control of mitochondrial apoptosis.. PLoS pathogens(IF=4.9). 2008. PMID:18516228. DOI: 10.1371/journal.ppat.1000018.
  • [22] Zukile Mbita;Rodney Hull;Zodwa Dlamini. Human immunodeficiency virus-1 (HIV-1)-mediated apoptosis: new therapeutic targets.. Viruses(IF=3.5). 2014. PMID:25196285. DOI: 10.3390/v6083181.
  • [23] Marie-Lise Gougeon. Apoptosis as an HIV strategy to escape immune attack.. Nature reviews. Immunology(IF=60.9). 2003. PMID:12766761. DOI: 10.1038/nri1087.
  • [24] Yu You;An-Chun Cheng;Ming-Shu Wang;Ren-Yong Jia;Kun-Feng Sun;Qiao Yang;Ying Wu;Dekang Zhu;Shun Chen;Ma-Feng Liu;Xin-Xin Zhao;Xiao-Yue Chen. The suppression of apoptosis by α-herpesvirus.. Cell death & disease(IF=9.6). 2017. PMID:28406478. DOI: 10.1038/cddis.2017.139.
  • [25] Chris A Benedict;Paula S Norris;Carl F Ware. To kill or be killed: viral evasion of apoptosis.. Nature immunology(IF=27.6). 2002. PMID:12407409. DOI: 10.1038/ni1102-1013.
  • [26] Ying Kai Chan;Michaela U Gack. Viral evasion of intracellular DNA and RNA sensing.. Nature reviews. Microbiology(IF=103.3). 2016. PMID:27174148. DOI: 10.1038/nrmicro.2016.45.
  • [27] Yangqi Yin;Xuyang Chi;Yandong Feng;Qinglian Jiang. The strategies and mechanisms of enteroviruses to evade innate immunity and the vaccine progress of enteroviruses.. Frontiers in cellular and infection microbiology(IF=4.8). 2025. PMID:40822588. DOI: 10.3389/fcimb.2025.1636104.
  • [28] Maroof Hasan;Nan Yan. Safeguard against DNA sensing: the role of TREX1 in HIV-1 infection and autoimmune diseases.. Frontiers in microbiology(IF=4.5). 2014. PMID:24817865. DOI: 10.3389/fmicb.2014.00193.
  • [29] Ling Wang;Dandan He;Naoko Satoh-Takayama;Chunfu Zheng;Junji Xing. Regulation of antiviral and antimicrobial innate immunity and immune evasion.. Cellular and molecular life sciences : CMLS(IF=6.2). 2025. PMID:40879755. DOI: 10.1007/s00018-025-05864-w.
  • [30] Junji Zhu;Cindy Chiang;Michaela U Gack. Viral evasion of the interferon response at a glance.. Journal of cell science(IF=3.6). 2023. PMID:37341132. DOI: 10.1242/jcs.260682.
  • [31] Herman W Favoreel;Gerlinde R Van de Walle;Hans J Nauwynck;Maurice B Pensaert. Virus complement evasion strategies.. The Journal of general virology(IF=4.3). 2003. PMID:12533696. DOI: 10.1099/vir.0.18709-0.
  • [32] Jack Mellors;Tom Tipton;Stephanie Longet;Miles Carroll. Viral Evasion of the Complement System and Its Importance for Vaccines and Therapeutics.. Frontiers in immunology(IF=5.9). 2020. PMID:32733480. DOI: 10.3389/fimmu.2020.01450.
  • [33] Bailey E Maloney;Krishani Dinali Perera;Danielle R D Saunders;Naemi Shadipeni;Sherry D Fleming. Interactions of viruses and the humoral innate immune response.. Clinical immunology (Orlando, Fla.)(IF=3.8). 2020. PMID:32028020. DOI: 10.1016/j.clim.2020.108351.
  • [34] Jacelyn Ms Loh;Haniyeh Aghababa;Thomas Proft. Eluding the immune system's frontline defense: Secreted complement evasion factors of pathogenic Gram-positive cocci.. Microbiological research(IF=6.9). 2023. PMID:37826985. DOI: 10.1016/j.micres.2023.127512.
  • [35] Ying Zhang;Jingyan Li;Qihan Li. Immune Evasion of Enteroviruses Under Innate Immune Monitoring.. Frontiers in microbiology(IF=4.5). 2018. PMID:30154774. DOI: 10.3389/fmicb.2018.01866.
  • [36] Adam John Ritchie;Fangping Cai;Nicola M G Smith;Sheri Chen;Hongshuo Song;Simon Brackenridge;Salim S Abdool Karim;Bette T Korber;Andrew J McMichael;Feng Gao;Nilu Goonetilleke. Recombination-mediated escape from primary CD8+ T cells in acute HIV-1 infection.. Retrovirology(IF=3.9). 2014. PMID:25212771. DOI: 10.1186/s12977-014-0069-9.
  • [37] Anouk Bc Schuren;Ana I Costa;Emmanuel Jhj Wiertz. Recent advances in viral evasion of the MHC Class I processing pathway.. Current opinion in immunology(IF=5.8). 2016. PMID:27065088. DOI: .
  • [38] Kehong Zheng;Xiaojun Zheng;Wei Yang. The Role of Metabolic Dysfunction in T-Cell Exhaustion During Chronic Viral Infection.. Frontiers in immunology(IF=5.9). 2022. PMID:35432304. DOI: 10.3389/fimmu.2022.843242.
  • [39] Robert Thimme;Marco Binder;Ralf Bartenschlager. Failure of innate and adaptive immune responses in controlling hepatitis C virus infection.. FEMS microbiology reviews(IF=12.3). 2012. PMID:22142141. DOI: 10.1111/j.1574-6976.2011.00319.x.
  • [40] C Brander;B D Walker. Modulation of host immune responses by clinically relevant human DNA and RNA viruses.. Current opinion in microbiology(IF=7.5). 2000. PMID:10972498. DOI: 10.1016/s1369-5274(00)00108-9.
  • [41] Mancini Nicasio;Giuseppe Sautto;Nicola Clementi;Roberta A Diotti;Elena Criscuolo;Matteo Castelli;Laura Solforosi;Massimo Clementi;Roberto Burioni. Neutralization interfering antibodies: a "novel" example of humoral immune dysfunction facilitating viral escape?. Viruses(IF=3.5). 2012. PMID:23170181. DOI: 10.3390/v4091731.
  • [42] Davide Angeletti;Jonathan W Yewdell. Understanding and Manipulating Viral Immunity: Antibody Immunodominance Enters Center Stage.. Trends in immunology(IF=13.9). 2018. PMID:29789196. DOI: 10.1016/j.it.2018.04.008.
  • [43] Isabel Fofana;Samira Fafi-Kremer;Patric Carolla;Catherine Fauvelle;Muhammad Nauman Zahid;Marine Turek;Laura Heydmann;Karine Cury;Juliette Hayer;Christophe Combet;François-Loïc Cosset;Thomas Pietschmann;Marie-Sophie Hiet;Ralf Bartenschlager;François Habersetzer;Michel Doffoël;Zhen-Yong Keck;Steven K H Foung;Mirjam B Zeisel;Françoise Stoll-Keller;Thomas F Baumert. Mutations that alter use of hepatitis C virus cell entry factors mediate escape from neutralizing antibodies.. Gastroenterology(IF=25.1). 2012. PMID:22503792. DOI: 10.1053/j.gastro.2012.04.006.
  • [44] Olga Tarasova;Anthi Petrou;Sergey M Ivanov;Athina Geronikaki;Vladimir Poroikov. Viral Factors in Modulation of Host Immune Response: A Route to Novel Antiviral Agents and New Therapeutic Approaches.. International journal of molecular sciences(IF=4.9). 2024. PMID:39273355. DOI: 10.3390/ijms25179408.
  • [45] Connie Zhao;Hongru Li;Talia H Swartz;Benjamin K Chen. The HIV Env Glycoprotein Conformational States on Cells and Viruses.. mBio(IF=4.7). 2022. PMID:35323042. DOI: 10.1128/mbio.01825-21.
  • [46] W Kamp;M B Berk;C J Visser;H S Nottet. Mechanisms of HIV-1 to escape from the host immune surveillance.. European journal of clinical investigation(IF=3.6). 2000. PMID:10964167. DOI: 10.1046/j.1365-2362.2000.00697.x.
  • [47] Zhongyue Fang;Wenling Jiang;Pei Liu;Ningshao Xia;Shaowei Li;Ying Gu. Targeting HIV-1 immune escape mechanisms: Key advances and challenges in HIV-1 vaccine design.. Microbiological research(IF=6.9). 2025. PMID:40460810. DOI: 10.1016/j.micres.2025.128229.
  • [48] Lu Lu;Fei Yu;Lan-Ying DU;Wei Xu;Shi-Bo Jiang. Tactics used by HIV-1 to evade host innate, adaptive, and intrinsic immunities.. Chinese medical journal(IF=7.3). 2013. PMID:23786957. DOI: .
  • [49] Angelique Hölzemer;Christina F Thobakgale;Camilo A Jimenez Cruz;Wilfredo F Garcia-Beltran;Jonathan M Carlson;Nienke H van Teijlingen;Jaclyn K Mann;Manjeetha Jaggernath;Seung-gu Kang;Christian Körner;Amy W Chung;Jamie L Schafer;David T Evans;Galit Alter;Bruce D Walker;Philip J Goulder;Mary Carrington;Pia Hartmann;Thomas Pertel;Ruhong Zhou;Thumbi Ndung'u;Marcus Altfeld. Selection of an HLA-C*03:04-Restricted HIV-1 p24 Gag Sequence Variant Is Associated with Viral Escape from KIR2DL3+ Natural Killer Cells: Data from an Observational Cohort in South Africa.. PLoS medicine(IF=9.9). 2015. PMID:26575988. DOI: 10.1371/journal.pmed.1001900.
  • [50] Teena Mohan;Santwana Bhatnagar;Dablu L Gupta;D N Rao. Current understanding of HIV-1 and T-cell adaptive immunity: progress to date.. Microbial pathogenesis(IF=3.5). 2014. PMID:24930593. DOI: .
  • [51] Hao Jiang;Si-Mei Shen;Jie Yin;Peng-Peng Zhang;Yi Shi. Influenza virus non‑structural protein 1 inhibits the production of interferon β of alveolar epithelial cells upon the infection of influenza A H1N1.. Molecular medicine reports(IF=3.5). 2017. PMID:28765967. DOI: 10.3892/mmr.2017.7138.
  • [52] Rachel Emily Levene;Marta Maria Gaglia. Host Shutoff in Influenza A Virus: Many Means to an End.. Viruses(IF=3.5). 2018. PMID:30189604. DOI: 10.3390/v10090475.
  • [53] Yotam Bar-On;Ariella Glasner;Tal Meningher;Hagit Achdout;Chamutal Gur;Dikla Lankry;Alon Vitenshtein;Adrienne F A Meyers;Michal Mandelboim;Ofer Mandelboim. Neuraminidase-mediated, NKp46-dependent immune-evasion mechanism of influenza viruses.. Cell reports(IF=6.9). 2013. PMID:23602571. DOI: 10.1016/j.celrep.2013.03.034.
  • [54] Qingsen Wang;Wenliang Pan;Song Wang;Chen Pan;Hongya Ning;Shile Huang;Shih-Hsin Chiu;Ji-Long Chen. Protein Tyrosine Phosphatase SHP2 Suppresses Host Innate Immunity against Influenza A Virus by Regulating EGFR-Mediated Signaling.. Journal of virology(IF=3.8). 2021. PMID:33361428. DOI: 10.1128/JVI.02001-20.
  • [55] Chengye Li;Tong Wang;Yuying Zhang;Fanhua Wei. Evasion mechanisms of the type I interferons responses by influenza A virus.. Critical reviews in microbiology(IF=5.1). 2020. PMID:32715811. DOI: 10.1080/1040841X.2020.1794791.
  • [56] Zhang Xu;Jung-Hyun Choi;David L Dai;Jun Luo;Reese Jalal Ladak;Qian Li;Yimeng Wang;Christine Zhang;Shane Wiebe;Alex C H Liu;Xiaozhuo Ran;Jiaqi Yang;Parisa Naeli;Aitor Garzia;Lele Zhou;Niaz Mahmood;Qiyun Deng;Mohamed Elaish;Rongtuan Lin;Lara K Mahal;Tom C Hobman;Jerry Pelletier;Tommy Alain;Silvia M Vidal;Thomas Duchaine;Mohammad T Mazhab-Jafari;Xiaojuan Mao;Seyed Mehdi Jafarnejad;Nahum Sonenberg. SARS-CoV-2 impairs interferon production via NSP2-induced repression of mRNA translation.. Proceedings of the National Academy of Sciences of the United States of America(IF=9.1). 2022. PMID:35878012. DOI: 10.1073/pnas.2204539119.
  • [57] Jordan A Hartmann;Marcella R Cardoso;Maria Cecilia Ramiro Talarico;Devin J Kenney;Madison R Leone;Dagny C Reese;Jacquelyn Turcinovic;Aoife K O'Connell;Hans P Gertje;Caitlin Marino;Pedro E Ojeda;Erich V De Paula;Fernanda A Orsi;Licio Augusto Velloso;Thomas R Cafiero;John H Connor;Alexander Ploss;Angelique Hoelzemer;Mary Carrington;Amy K Barczak;Nicholas A Crossland;Florian Douam;Julie Boucau;Wilfredo F Garcia-Beltran. Evasion of NKG2D-mediated cytotoxic immunity by sarbecoviruses.. Cell(IF=42.5). 2024. PMID:38653235. DOI: 10.1016/j.cell.2024.03.026.
  • [58] Qian Wang;Xing Wu;Qunying Mao;Fan Gao;Mingchen Liu;Ziyang Song;Lianlian Bian;Zhenglun Liang. How SARS-CoV-2 dodges immune surveillance and facilitates infection: an analytical review.. Expert review of anti-infective therapy(IF=3.8). 2022. PMID:35574688. DOI: 10.1080/14787210.2022.2078307.
  • [59] Sameer-Ul-Salam Mattoo;Seong-Jun Kim;Dae-Gyun Ahn;Jinjong Myoung. Escape and Over-Activation of Innate Immune Responses by SARS-CoV-2: Two Faces of a Coin.. Viruses(IF=3.5). 2022. PMID:35336937. DOI: 10.3390/v14030530.
  • [60] Yu-Zhi Fu;Su-Yun Wang;Zhou-Qin Zheng; Yi Huang;Wei-Wei Li;Zhi-Sheng Xu;Yan-Yi Wang. SARS-CoV-2 membrane glycoprotein M antagonizes the MAVS-mediated innate antiviral response.. Cellular & molecular immunology(IF=19.8). 2021. PMID:33110251. DOI: 10.1038/s41423-020-00571-x.
  • [61] Bingqing Xia;Xiaoyan Pan;Rong-Hua Luo;Xurui Shen;Shuangqu Li;Yi Wang;Xiaoli Zuo;Yan Wu;Yingqi Guo;Gengfu Xiao;Qiguang Li;Xin-Yan Long;Xiao-Yan He;Hong-Yi Zheng;Ying Lu;Wei Pang;Yong-Tang Zheng;Jia Li;Lei-Ke Zhang;Zhaobing Gao. Extracellular vesicles mediate antibody-resistant transmission of SARS-CoV-2.. Cell discovery(IF=12.5). 2023. PMID:36609376. DOI: 10.1038/s41421-022-00510-2.
  • [62] Douglas Jie Wen Tay;Zhe Zhang Ryan Lew;Justin Jang Hann Chu;Kai Sen Tan. Uncovering Novel Viral Innate Immune Evasion Strategies: What Has SARS-CoV-2 Taught Us?. Frontiers in microbiology(IF=4.5). 2022. PMID:35401477. DOI: 10.3389/fmicb.2022.844447.
  • [63] Chiranjib Chakraborty;Ashish Ranjan Sharma;Manojit Bhattacharya;Sang-Soo Lee. A Detailed Overview of Immune Escape, Antibody Escape, Partial Vaccine Escape of SARS-CoV-2 and Their Emerging Variants With Escape Mutations.. Frontiers in immunology(IF=5.9). 2022. PMID:35222380. DOI: 10.3389/fimmu.2022.801522.
  • [64] Wenjing Gu;Hui Gan;Yu Ma;Lina Xu;Zhangkai J Cheng;Bizhou Li;Xinxing Zhang;Wujun Jiang;Jinlv Sun;Baoqing Sun;Chuangli Hao. The molecular mechanism of SARS-CoV-2 evading host antiviral innate immunity.. Virology journal(IF=3.8). 2022. PMID:35305698. DOI: 10.1186/s12985-022-01783-5.

MaltSci Intelligent Research Services

Search for more papers on MaltSci.com

viral immune evasion · host immune system · vaccine development


© 2025 MaltSci